Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Mol Immunol ; 21(4): 374-392, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38383773

RESUMEN

CD4+ T cells can "help" or "license" conventional type 1 dendritic cells (cDC1s) to induce CD8+ cytotoxic T lymphocyte (CTL) anticancer responses, as proven in mouse models. We recently identified cDC1s with a transcriptomic imprint of CD4+ T-cell help, specifically in T-cell-infiltrated human cancers, and these cells were associated with a good prognosis and response to PD-1-targeting immunotherapy. Here, we delineate the mechanism of cDC1 licensing by CD4+ T cells in humans. Activated CD4+ T cells produce IFNß via the STING pathway, which promotes MHC-I antigen (cross-)presentation by cDC1s and thereby improves their ability to induce CTL anticancer responses. In cooperation with CD40 ligand (L), IFNß also optimizes the costimulatory and other functions of cDC1s required for CTL response induction. IFN-I-producing CD4+ T cells are present in diverse T-cell-infiltrated cancers and likely deliver "help" signals to CTLs locally, according to their transcriptomic profile and colocalization with "helped/licensed" cDCs and tumor-reactive CD8+ T cells. In agreement with this scenario, the presence of IFN-I-producing CD4+ T cells in the TME is associated with overall survival and the response to PD-1 checkpoint blockade in cancer patients.


Asunto(s)
Neoplasias , Linfocitos T Citotóxicos , Ratones , Animales , Humanos , Linfocitos T CD8-positivos , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T CD4-Positivos , Células Dendríticas
2.
EMBO J ; 41(6): e109760, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35156720

RESUMEN

RNA editing by the adenosine deaminase ADAR1 prevents innate immune responses to endogenous RNAs. In ADAR1-deficient cells, unedited self RNAs form base-paired structures that resemble viral RNAs and inadvertently activate the cytosolic RIG-I-like receptor (RLR) MDA5, leading to an antiviral type I interferon (IFN) response. Mutations in ADAR1 cause Aicardi-Goutières Syndrome (AGS), an autoinflammatory syndrome characterized by chronic type I IFN production. Conversely, ADAR1 loss and the consequent type I IFN production restricts tumor growth and potentiates the activity of some chemotherapeutics. Here, we show that another RIG-I-like receptor, LGP2, also has an essential role in the induction of a type I IFN response in ADAR1-deficient human cells. This requires the canonical function of LGP2 as an RNA sensor and facilitator of MDA5-dependent signaling. Furthermore, we show that the sensitivity of tumor cells to ADAR1 loss requires LGP2 expression. Finally, type I IFN induction in tumor cells depleted of ADAR1 and treated with some chemotherapeutics fully depends on LGP2 expression. These findings highlight a central role for LGP2 in self RNA sensing with important clinical implications.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , Malformaciones del Sistema Nervioso , ARN Helicasas/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/genética , Humanos , Malformaciones del Sistema Nervioso/genética , Edición de ARN , ARN Bicatenario
3.
Nat Commun ; 11(1): 5387, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33144593

RESUMEN

The Human Silencing Hub (HUSH) complex is necessary for epigenetic repression of LINE-1 elements. We show that HUSH-depletion in human cell lines and primary fibroblasts leads to induction of interferon-stimulated genes (ISGs) through JAK/STAT signaling. This effect is mainly attributed to MDA5 and RIG-I sensing of double-stranded RNAs (dsRNAs). This coincides with upregulation of primate-conserved LINE-1s, as well as increased expression of full-length hominid-specific LINE-1s that produce bidirectional RNAs, which may form dsRNA. Notably, LTRs nearby ISGs are derepressed likely rendering these genes more responsive to interferon. LINE-1 shRNAs can abrogate the HUSH-dependent response, while overexpression of an engineered LINE-1 construct activates interferon signaling. Finally, we show that the HUSH component, MPP8 is frequently downregulated in diverse cancers and that its depletion leads to DNA damage. These results suggest that LINE-1s may drive physiological or autoinflammatory responses through dsRNA sensing and gene-regulatory roles and are controlled by the HUSH complex.


Asunto(s)
Epigénesis Genética/fisiología , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen/fisiología , Interferón Tipo I/metabolismo , Elementos de Nucleótido Esparcido Largo/fisiología , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Daño del ADN , Regulación hacia Abajo , Técnicas de Inactivación de Genes , Células HEK293 , Células HeLa , Humanos , Inflamación , Helicasa Inducida por Interferón IFIH1/metabolismo , Elementos de Nucleótido Esparcido Largo/genética , Fosfoproteínas/metabolismo , ARN Bicatenario , Receptores Inmunológicos , Análisis de Secuencia de ARN , Transducción de Señal
4.
J Immunol ; 205(4): 883-891, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32769143

RESUMEN

The innate immune system uses pattern recognition receptors to survey the intracellular and extracellular environment for signs of infection. Viral infection is detected through the presence of viral nucleic acids in infected cells. Pattern recognition receptor activation by viral nucleic acids induces the expression and secretion of type I IFNs (IFN-Is), important mediators of antiviral immunity. RIG-I-like receptors (RLRs) are RNA sensors that detect viral RNA in the cytosol and induce an IFN-I response. Viral RNAs contain features that set them apart from host RNAs, allowing RLRs to discriminate between cellular/self and viral/non-self RNA. The notion emerged that self RNAs can also engage RLRs and modulate the IFN-I response, indicating that the distinction between self and non-self RNA is not watertight. We review how self RNAs regulate RLR activation and the IFN-I response during viral infection and how recognition of self RNAs by RLRs is implicated in autoinflammatory disorders and cancer.


Asunto(s)
Inflamación/inmunología , Interferón Tipo I/inmunología , ARN Viral/inmunología , Receptores de Interferón/inmunología , Virosis/inmunología , Animales , Humanos , Receptores de Reconocimiento de Patrones/inmunología
5.
Cytokine Growth Factor Rev ; 55: 1-14, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32563552

RESUMEN

Rapid detection of microbes is crucial for eliciting an effective immune response. Innate immune receptors survey the intracellular and extracellular environment for signs of a microbial infection. When they detect a pathogen-associated molecular pattern (PAMP), such as viral DNA, they alarm the cell about the ongoing infection. The central signaling hub in sensing of viral DNA is the stimulator of interferon genes (STING). Upon activation, STING induces downstream signaling events that ultimately result in the production of type I interferons (IFN I), important cytokines in antimicrobial defense, in particular towards viruses. In this review, we describe the molecular features of STING, including its upstream sensors and ligands, its sequence and structural conservation, common polymorphisms, and its localization. We further highlight how STING activation requires a careful balance: its activity is essential for antiviral defense, but unwanted activation through mutations or accidental recognition of self-derived DNA causes autoinflammatory diseases. Several mechanisms, such as post-translational modifications, ensure this balance by fine-tuning STING activation. Finally, we discuss how viruses evade detection of their genomes by either exploiting cells that lack a functional DNA sensing pathway as a niche or by interfering with STING activation through viral evasion molecules. Insight into STING's exact mechanisms in health and disease will guide the development of novel clinical interventions for microbial infections, autoinflammatory diseases, and beyond.


Asunto(s)
Inmunidad Innata , Inflamación , Interferón Tipo I , Proteínas de la Membrana , Citocinas , Inflamación/inmunología , Proteínas de la Membrana/inmunología , Transducción de Señal
6.
EMBO J ; 38(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30872283

RESUMEN

To protect against the harmful consequences of viral infections, organisms are equipped with sophisticated antiviral mechanisms, including cell-intrinsic means to restrict viral replication and propagation. Plant and invertebrate cells utilise mostly RNA interference (RNAi), an RNA-based mechanism, for cell-intrinsic immunity to viruses while vertebrates rely on the protein-based interferon (IFN)-driven innate immune system for the same purpose. The RNAi machinery is conserved in vertebrate cells, yet whether antiviral RNAi is still active in mammals and functionally relevant to mammalian antiviral defence is intensely debated. Here, we discuss cellular and viral factors that impact on antiviral RNAi and the contexts in which this system might be at play in mammalian resistance to viral infection.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Mamíferos/inmunología , Interferencia de ARN , ARN Viral/genética , Virosis/inmunología , Virus/inmunología , Animales , Antivirales/administración & dosificación , Interacciones Huésped-Patógeno/genética , Mamíferos/genética , Mamíferos/virología , Virosis/genética , Virosis/virología , Replicación Viral , Virus/aislamiento & purificación
7.
J Immunol ; 201(2): 604-614, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29891555

RESUMEN

IFN-stimulated gene (ISG) 15 is a ubiquitin-like protein induced after type I IFN stimulation. There is a dearth of in vivo models to study free unconjugated ISG15 function. We found that free ISG15 enhances the production of IFN-γ and IL-1ß during murine infection with Toxoplasma gondii In our model, ISG15 is induced in a type I IFN-dependent fashion and released into the serum. Increased ISG15 levels are dependent on an actively invading and replicating parasite. Two cysteine residues in the hinge domain are necessary determinants for ISG15 to induce increased cytokine levels during infection. Increased ISG15 is concurrent with an influx of IL-1ß-producing CD8α+ dendritic cells to the site of infection. In this article, we present Toxoplasma infection as a novel in vivo murine model to study the immunomodulatory properties of free ISG15 and uniquely link it to IL-1ß production by CD8α+ dendritic cells driven by two cysteines in the hinge region of the protein.


Asunto(s)
Citocinas/metabolismo , Células Dendríticas/inmunología , Interleucina-1beta/metabolismo , Toxoplasma/fisiología , Toxoplasmosis/inmunología , Animales , Antígenos CD8/metabolismo , Movimiento Celular , Células Cultivadas , Cisteína/genética , Citocinas/genética , Modelos Animales de Enfermedad , Inmunomodulación , Interferón Tipo I/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Conformación Proteica , Ubiquitinas/genética , Ubiquitinas/metabolismo
8.
Nat Commun ; 9(1): 1820, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29739942

RESUMEN

RIG-I is a viral RNA sensor that induces the production of type I interferon (IFN) in response to infection with a variety of viruses. Modification of RIG-I with K63-linked poly-ubiquitin chains, synthesised by TRIM25, is crucial for activation of the RIG-I/MAVS signalling pathway. TRIM25 activity is targeted by influenza A virus non-structural protein 1 (NS1) to suppress IFN production and prevent an efficient host immune response. Here we present structures of the human TRIM25 coiled-coil-PRYSPRY module and of complexes between the TRIM25 coiled-coil domain and NS1. These structures show that binding of NS1 interferes with the correct positioning of the PRYSPRY domain of TRIM25 required for substrate ubiquitination and provide a mechanistic explanation for how NS1 suppresses RIG-I ubiquitination and hence downstream signalling. In contrast, the formation of unanchored K63-linked poly-ubiquitin chains is unchanged by NS1 binding, indicating that RING dimerisation of TRIM25 is not affected by NS1.


Asunto(s)
Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/inmunología , Proteínas de Motivos Tripartitos/antagonistas & inhibidores , Proteínas de Motivos Tripartitos/inmunología , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/inmunología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/inmunología , Secuencia de Aminoácidos , Células Cultivadas , Proteína 58 DEAD Box/inmunología , Células HEK293 , Humanos , Interferones/biosíntesis , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , ARN Viral/inmunología , Receptores Inmunológicos , Transducción de Señal , Factores de Transcripción/química , Proteínas de Motivos Tripartitos/química , Ubiquitina-Proteína Ligasas/química , Ubiquitinación
9.
EMBO J ; 37(4)2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29351913

RESUMEN

In vertebrates, the presence of viral RNA in the cytosol is sensed by members of the RIG-I-like receptor (RLR) family, which signal to induce production of type I interferons (IFN). These key antiviral cytokines act in a paracrine and autocrine manner to induce hundreds of interferon-stimulated genes (ISGs), whose protein products restrict viral entry, replication and budding. ISGs include the RLRs themselves: RIG-I, MDA5 and, the least-studied family member, LGP2. In contrast, the IFN system is absent in plants and invertebrates, which defend themselves from viral intruders using RNA interference (RNAi). In RNAi, the endoribonuclease Dicer cleaves virus-derived double-stranded RNA (dsRNA) into small interfering RNAs (siRNAs) that target complementary viral RNA for cleavage. Interestingly, the RNAi machinery is conserved in mammals, and we have recently demonstrated that it is able to participate in mammalian antiviral defence in conditions in which the IFN system is suppressed. In contrast, when the IFN system is active, one or more ISGs act to mask or suppress antiviral RNAi. Here, we demonstrate that LGP2 constitutes one of the ISGs that can inhibit antiviral RNAi in mammals. We show that LGP2 associates with Dicer and inhibits cleavage of dsRNA into siRNAs both in vitro and in cells. Further, we show that in differentiated cells lacking components of the IFN response, ectopic expression of LGP2 interferes with RNAi-dependent suppression of gene expression. Conversely, genetic loss of LGP2 uncovers dsRNA-mediated RNAi albeit less strongly than complete loss of the IFN system. Thus, the inefficiency of RNAi as a mechanism of antiviral defence in mammalian somatic cells can be in part attributed to Dicer inhibition by LGP2 induced by type I IFNs. LGP2-mediated antagonism of dsRNA-mediated RNAi may help ensure that viral dsRNA substrates are preserved in order to serve as targets of antiviral ISG proteins.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Interferón Tipo I/metabolismo , ARN Helicasas/metabolismo , Interferencia de ARN , Virus ARN/fisiología , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/genética , Ribonucleasa III/metabolismo , ARN Helicasas DEAD-box/genética , Regulación de la Expresión Génica , Células HeLa , Humanos , ARN Helicasas/genética , ARN Bicatenario/genética , ARN Viral/genética , Ribonucleasa III/genética , Transducción de Señal
10.
Cell Rep ; 20(3): 529-537, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723558

RESUMEN

Conventional dendritic cells (cDCs) are thought to descend from a DC precursor downstream of the common myeloid progenitor (CMP). However, a mouse lymphoid-primed multipotent progenitor has been shown to generate cDCs following a DC-specific developmental pathway independent of monocyte and granulocyte poiesis. Similarly, here we show that, in humans, a large fraction of multipotent lymphoid early progenitors (MLPs) gives rise to cDCs, in particular the subset known as cDC1, identified by co-expression of DNGR-1 (CLEC9A) and CD141 (BDCA-3). Single-cell analysis indicates that over one-third of MLPs have the potential to efficiently generate cDCs. cDC1s generated from CMPs or MLPs do not exhibit differences in transcriptome or phenotype. These results demonstrate an early imprinting of the cDC lineage in human hematopoiesis and highlight the plasticity of developmental pathways giving rise to human DCs.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Diferenciación Celular , Células Dendríticas/metabolismo , Células Progenitoras Linfoides/metabolismo , Células Madre Multipotentes/metabolismo , Animales , Células Dendríticas/citología , Humanos , Células Progenitoras Linfoides/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células Madre Multipotentes/citología
11.
EMBO J ; 35(23): 2505-2518, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27815315

RESUMEN

RNA interference (RNAi) elicited by long double-stranded (ds) or base-paired viral RNA constitutes the major mechanism of antiviral defence in plants and invertebrates. In contrast, it is controversial whether it acts in chordates. Rather, in vertebrates, viral RNAs induce a distinct defence system known as the interferon (IFN) response. Here, we tested the possibility that the IFN response masks or inhibits antiviral RNAi in mammalian cells. Consistent with that notion, we find that sequence-specific gene silencing can be triggered by long dsRNAs in differentiated mouse cells rendered deficient in components of the IFN pathway. This unveiled response is dependent on the canonical RNAi machinery and is lost upon treatment of IFN-responsive cells with type I IFN Notably, transfection with long dsRNA specifically vaccinates IFN-deficient cells against infection with viruses bearing a homologous sequence. Thus, our data reveal that RNAi constitutes an ancient antiviral strategy conserved from plants to mammals that precedes but has not been superseded by vertebrate evolution of the IFN system.


Asunto(s)
Regulación de la Expresión Génica , Interferencia de ARN , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Animales , Células Cultivadas , Inmunidad Innata , Interferón Tipo I/antagonistas & inhibidores , Ratones , Virus ARN/inmunología
13.
Eur J Immunol ; 45(12): 3386-403, 2015 12.
Artículo en Inglés | MEDLINE | ID: mdl-26457795

RESUMEN

IFN-α/ß allow cells to fight virus infection by inducing the expression of many genes that encode effectors of antiviral defense. One of these, the Ski2-like DExH-box helicase DDX60, was recently implicated in resistance of human cells to hepatitis C virus, as well as in induction of IFN-α/ß by retinoic acid inducible gene 1-like receptors (RLRs) that detect the presence of RNA viruses in a cell-intrinsic manner. Here, we sought to investigate the role of DDX60 in IFN-α/ß induction and in resistance to virus infection. Analysis of fibroblasts and myeloid cells from Ddx60-deficient mice revealed no impairment in IFN-α/ß production in response to RLR agonists, RNA viruses, or other stimuli. Moreover, overexpression of DDX60 did not potentiate IFN induction and DDX60 did not interact with RLRs or capture RLR agonists from virally infected cells. We also failed to identify any impairment in Ddx60-deficient murine cells or mice in resistance to infection with influenza A virus, encephalomyocarditis virus, Sindbis virus, vaccinia virus, or herpes simplex virus-1. These results put in question the reported role of DDX60 as a broad-acting positive regulator of RLR responses and hint at the possibility that it may function as a restriction factor highly specific for a particular virus or class of viruses.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Interferón Tipo I/biosíntesis , Virosis/inmunología , Animales , Línea Celular , Citocinas/biosíntesis , Humanos , Ratones , Receptores Toll-Like/fisiología
14.
Cell ; 162(6): 1257-70, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26343581

RESUMEN

The mechanisms by which melanoma and other cancer cells evade anti-tumor immunity remain incompletely understood. Here, we show that the growth of tumors formed by mutant Braf(V600E) mouse melanoma cells in an immunocompetent host requires their production of prostaglandin E2, which suppresses immunity and fuels tumor-promoting inflammation. Genetic ablation of cyclooxygenases (COX) or prostaglandin E synthases in Braf(V600E) mouse melanoma cells, as well as in Nras(G12D) melanoma or in breast or colorectal cancer cells, renders them susceptible to immune control and provokes a shift in the tumor inflammatory profile toward classic anti-cancer immune pathways. This mouse COX-dependent inflammatory signature is remarkably conserved in human cutaneous melanoma biopsies, arguing for COX activity as a driver of immune suppression across species. Pre-clinical data demonstrate that inhibition of COX synergizes with anti-PD-1 blockade in inducing eradication of tumors, implying that COX inhibitors could be useful adjuvants for immune-based therapies in cancer patients.


Asunto(s)
Neoplasias/inmunología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Escape del Tumor , Inmunidad Adaptativa , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD/inmunología , Aspirina/administración & dosificación , Línea Celular Tumoral , Células Dendríticas/inmunología , Humanos , Inmunidad Innata , Inmunoterapia , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Cadenas alfa de Integrinas/inmunología , Interferones/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Ratones , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Prostaglandinas/inmunología , Proteínas Proto-Oncogénicas B-raf/metabolismo
15.
J Immunol Methods ; 423: 120-30, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25967946

RESUMEN

Calcium ions (Ca(2+)) are a ubiquitous transducer of cellular signals controlling key processes such as proliferation, differentiation, secretion and metabolism. In the context of T cells, stimulation through the T cell receptor has been shown to induce the release of Ca(2+) from intracellular stores. This sudden elevation within the cytoplasm triggers the opening of ion channels in the plasma membrane allowing an influx of extracellular Ca(2+) that in turn activates key molecules such as calcineurin. This cascade ultimately results in gene transcription and changes in the cellular state. Traditional methods for measuring Ca(2+) include spectrophotometry, conventional flow cytometry (CFC) and live cell imaging techniques. While each method has strengths and weaknesses, none can offer a detailed picture of Ca(2+) mobilisation in response to various agonists. Here we report an Imaging Flow Cytometry (IFC)-based method that combines the throughput and statistical rigour of CFC with the spatial information of a microscope. By co-staining cells with Ca(2+) indicators and organelle-specific dyes we can address the spatiotemporal patterns of Ca(2+) flux in Jurkat cells after stimulation with anti-CD3. The multispectral, high-throughput nature of IFC means that the organelle co-staining functions to direct the measurement of Ca(2+) indicator fluorescence to either the endoplasmic reticulum (ER) or the mitochondrial compartments without the need to treat cells with detergents such as digitonin to eliminate cytoplasmic background. We have used this system to look at the cellular localisation of Ca(2+) after stimulating cells with CD3, thapsigargin or ionomycin in the presence or absence of extracellular Ca(2+). Our data suggest that there is a dynamic interplay between the ER and mitochondrial compartments and that mitochondria act as a sink for both intracellular and extracellular derived Ca(2+). Moreover, by generating an NFAT-GFP expressing Jurkat line, we were able to combine mitochondrial Ca(2+) measurements with nuclear translocation. In conclusion, this method enables the high throughput study of spatiotemporal patterns of Ca2(+) signals in T cells responding to different stimuli.


Asunto(s)
Calcio/metabolismo , Activación de Linfocitos/fisiología , Linfocitos T/metabolismo , Linfocitos T/fisiología , Complejo CD3/metabolismo , Línea Celular Tumoral , Citoplasma/metabolismo , Retículo Endoplásmico/metabolismo , Citometría de Flujo/métodos , Humanos , Citometría de Imagen/métodos , Células Jurkat , Microscopía/métodos , Mitocondrias/metabolismo , Análisis Espacial , Análisis Espacio-Temporal
16.
Nature ; 514(7522): 372-375, 2014 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-25119032

RESUMEN

Mammalian cells possess mechanisms to detect and defend themselves from invading viruses. In the cytosol, the RIG-I-like receptors (RLRs), RIG-I (retinoic acid-inducible gene I; encoded by DDX58) and MDA5 (melanoma differentiation-associated gene 5; encoded by IFIH1) sense atypical RNAs associated with virus infection. Detection triggers a signalling cascade via the adaptor MAVS that culminates in the production of type I interferons (IFN-α and ß; hereafter IFN), which are key antiviral cytokines. RIG-I and MDA5 are activated by distinct viral RNA structures and much evidence indicates that RIG-I responds to RNAs bearing a triphosphate (ppp) moiety in conjunction with a blunt-ended, base-paired region at the 5'-end (reviewed in refs 1, 2, 3). Here we show that RIG-I also mediates antiviral responses to RNAs bearing 5'-diphosphates (5'pp). Genomes from mammalian reoviruses with 5'pp termini, 5'pp-RNA isolated from yeast L-A virus, and base-paired 5'pp-RNAs made by in vitro transcription or chemical synthesis, all bind to RIG-I and serve as RIG-I agonists. Furthermore, a RIG-I-dependent response to 5'pp-RNA is essential for controlling reovirus infection in cultured cells and in mice. Thus, the minimal determinant for RIG-I recognition is a base-paired RNA with 5'pp. Such RNAs are found in some viruses but not in uninfected cells, indicating that recognition of 5'pp-RNA, like that of 5'ppp-RNA, acts as a powerful means of self/non-self discrimination by the innate immune system.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Difosfatos/metabolismo , Inmunidad Innata , ARN Viral/química , ARN Viral/metabolismo , Reoviridae/genética , Reoviridae/inmunología , Animales , Emparejamiento Base , Secuencia de Bases , Proteína 58 DEAD Box , Femenino , Genoma Viral/genética , Masculino , Ratones , ARN Viral/genética , Reoviridae/fisiología
17.
Annu Rev Biochem ; 81: 323-57, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22404627

RESUMEN

The eukaryotic ubiquitin family encompasses nearly 20 proteins that are involved in the posttranslational modification of various macromolecules. The ubiquitin-like proteins (UBLs) that are part of this family adopt the ß-grasp fold that is characteristic of its founding member ubiquitin (Ub). Although structurally related, UBLs regulate a strikingly diverse set of cellular processes, including nuclear transport, proteolysis, translation, autophagy, and antiviral pathways. New UBL substrates continue to be identified and further expand the functional diversity of UBL pathways in cellular homeostasis and physiology. Here, we review recent findings on such novel substrates, mechanisms, and functions of UBLs.


Asunto(s)
Ubiquitinas/metabolismo , Secuencias de Aminoácidos , Animales , Fenómenos Fisiológicos Celulares , Humanos , Procesamiento Proteico-Postraduccional , Sumoilación , Ubiquitinas/química , Levaduras/metabolismo
18.
Proc Natl Acad Sci U S A ; 108(22): 9238-43, 2011 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-21562218

RESUMEN

Type I strains of Helicobacter pylori (Hp) possess a pathogenicity island, cag, that encodes the effector protein cytotoxin-associated gene A (CagA) and a type four secretion system. After translocation into the host cell, CagA affects cell shape, increases cell motility, abrogates junctional activity, and promotes an epithelial to mesenchymal transition-like phenotype. Transgenic expression of CagA enhances gastrointestinal and intestinal carcinomas as well as myeloid and B-cell lymphomas in mice, but the mechanism of the induced cancer formation is not fully understood. Here, we show that CagA subverts the tumor suppressor function of apoptosis-stimulating protein of p53 (ASPP2). Delivery of CagA inside the host results in its association with ASPP2. After this interaction, ASPP2 recruits its natural target p53 and inhibits its apoptotic function. CagA leads to enhanced degradation of p53 and thereby, down-regulates its activity in an ASPP2-dependent manner. Finally, Hp-infected cells treated with the p53-activating drug Doxorubicin are more resistant to apoptosis than uninfected cells, an effect that requires ASPP2. The interaction between CagA and ASPP2 and the consequent degradation of p53 are examples of a bacterial protein that subverts the p53 tumor suppressor pathway in a manner similar to DNA tumor viruses. This finding may contribute to the understanding of the increased risk of gastric cancer in patients infected with Hp CagA+ strains.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Proteínas Bacterianas/metabolismo , Regulación de la Expresión Génica , Helicobacter pylori/metabolismo , Secuencia de Aminoácidos , ADN/metabolismo , Doxorrubicina/farmacología , Genes Supresores de Tumor , Humanos , Datos de Secuencia Molecular , Fenotipo , Riesgo , Neoplasias Gástricas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
19.
PLoS Biol ; 8(3): e1000605, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21468303

RESUMEN

Ubiquitin-dependent processes control much of cellular physiology. We show that expression of a highly active, Epstein-Barr virus-derived deubiquitylating enzyme (EBV-DUB) blocks proteasomal degradation of cytosolic and ER-derived proteins by preemptive removal of ubiquitin from proteasome substrates, a treatment less toxic than the use of proteasome inhibitors. Recognition of misfolded proteins in the ER lumen, their dislocation to the cytosol, and degradation are usually tightly coupled but can be uncoupled by the EBV-DUB: a misfolded glycoprotein that originates in the ER accumulates in association with cytosolic chaperones as a deglycosylated intermediate. Our data underscore the necessity of a DUB activity for completion of the dislocation reaction and provide a new means of inhibition of proteasomal proteolysis with reduced cytotoxicity.


Asunto(s)
Herpesvirus Humano 4/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Proteínas Virales/metabolismo , Biocatálisis , Línea Celular , Retículo Endoplásmico/metabolismo , Glicoproteínas/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Pliegue de Proteína , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Especificidad por Sustrato
20.
Proc Natl Acad Sci U S A ; 108(5): 1763-70, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21209336

RESUMEN

The ubiquitin (Ub)-related modifier Urm1 functions as a sulfur carrier in tRNA thiolation by means of a mechanism that requires the formation of a thiocarboxylate at the C-terminal glycine residue of Urm1. However, whether Urm1 plays an additional role as a Ub-like protein modifier remains unclear. Here, we show that Urm1 is conjugated to lysine residues of target proteins and that oxidative stress enhances protein urmylation in both Saccharomyces cerevisiae and mammalian cells. Similar to ubiquitylation, urmylation involves a thioester intermediate and results in the formation of a covalent peptide bond between Urm1 and its substrates. In contrast to modification by canonical Ub-like modifiers, however, conjugation of Urm1 involves a C-terminal thiocarboxylate of the modifier. We have confirmed that the peroxiredoxin Ahp1 is such a substrate in S. cerevisiae and found that Urm1 targets a specific lysine residue of Ahp1 in vivo. In addition, we have identified several unique substrates in mammalian cells and show that Urm1 targets at least two pathways on oxidant treatment. First, Urm1 is appended to lysine residues of three components that function in its own pathway (i.e., MOCS3, ATPBD3, and CTU2). Second, Urm1 is conjugated to the nucleocytoplasmic shuttling factor cellular apoptosis susceptibility protein. Thus, Urm1 has a conserved dual role by integrating the functions of prokaryotic sulfur carriers with those of eukaryotic protein modifiers of the Ub family.


Asunto(s)
Lisina/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Ubiquitinas/fisiología , Cromatografía Liquida , Diamida/metabolismo , Electroforesis en Gel de Poliacrilamida , Humanos , Estrés Oxidativo , Proteómica , Proteínas de Saccharomyces cerevisiae/metabolismo , Espectrometría de Masas en Tándem , Ubiquitinación , Ubiquitinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...